Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1868(9): 166428, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35533906

ABSTRACT

Aberrant DNA hypermethylation is associated with oral carcinogenesis. Procaine, a local anesthetic, is a DNA methyltransferase (DNMT) inhibitor that activates anticancer mechanisms. However, its effect on silenced tumor suppressor gene (TSG) activation and its biological role in oral squamous cell carcinoma (OSCC) remain unknown. Here, we report procaine inhibited DNA methylation by suppressing DNMT activity and increased the expression of PAX9, a differentiation gene in OSCC cells. Interestingly, the reactivation of PAX9 by procaine found to inhibit cell growth and trigger apoptosis in OSCC in vitro and in vivo. Likely, the enhanced PAX9 expression after exposure to procaine controls stemness and differentiation through the autophagy-dependent pathway in OSCC cells. PAX9 inhibition abrogated procaine-induced apoptosis, autophagy, and inhibition of stemness. In OSCC cells, procaine improved anticancer drug sensitivity through PAX9, and its deficiency significantly blunted the anticancer drug sensitivity mediated by procaine. Additionally, NRF2 activation by procaine facilitated the antitumor response of PAX9, and pharmacological inhibition of NRF2 by ML385 reduced death and prevented the decrease in the orosphere-forming potential of OSCC cells. Furthermore, procaine promoted antitumor activity in FaDu xenografts in athymic nude mice, and immunohistochemistry data showed that PAX9 expression was significantly enhanced in the procaine group compared to the vehicle control. In conclusion, PAX9 reactivation in response to DNMT inhibition could trigger a potent antitumor mechanism to provide a new therapeutic strategy for OSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , DNA , Humans , Methyltransferases , Mice , Mice, Nude , Mouth Neoplasms/drug therapy , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , NF-E2-Related Factor 2 , PAX9 Transcription Factor/genetics , PAX9 Transcription Factor/metabolism , Procaine/therapeutic use , Squamous Cell Carcinoma of Head and Neck
2.
Semin Cancer Biol ; 83: 399-412, 2022 08.
Article in English | MEDLINE | ID: mdl-33039557

ABSTRACT

Tumour-promoting inflammation is a critical hallmark in cancer development, and inflammasomes are well-known regulators of inflammatory processes within the tumour microenvironment. Different inflammasome components along with the adaptor, apoptosis-associated speck-like protein containing caspase activation and recruitment domain (ASC), and the effector, caspase-1, have a significant influence on tumorigenesis but in a tissue-specific and stage-dependent manner. The downstream products of inflammasome activation, that is the proinflammatory cytokines such as IL-1ß and IL-18, regulate tissue homeostasis and induce antitumour immune responses, but in contrast, they can also favour cancer growth and proliferation by directing various oncogenic signalling pathways in cancer cells. Moreover, different epigenetic mechanisms, including DNA methylation, histone modification and noncoding RNAs, control inflammasomes and their components by regulating gene expression during cancer progression. Furthermore, autophagy, a master controller of cellular homeostasis, targets inflammasome-induced carcinogenesis by maintaining cellular homeostasis and removing potential cancer risk factors that promote inflammasome activation in support of tumorigenesis. Here, in this review, we summarize the effect of inflammasome activation in cancers and discuss the role of epigenetic and autophagic regulatory mechanisms in controlling inflammasomes. A proper understanding of the interactions among these key processes will be useful for developing novel therapeutic regimens for targeting inflammasomes in cancer.


Subject(s)
Inflammasomes , Neoplasms , Autophagy/genetics , Carcinogenesis/genetics , Epigenesis, Genetic , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Neoplasms/genetics , Tumor Microenvironment/genetics
3.
Stem Cell Rev Rep ; 18(1): 198-213, 2022 01.
Article in English | MEDLINE | ID: mdl-34355273

ABSTRACT

Cancer stem cells (CSCs) are rare populations of malignant cells with stem cell-like features of self-renewal, uninterrupted differentiation, tumorigenicity, and resistance to conventional therapeutic agents, and these cells have a decisive role in treatment failure and tumor relapse. The self-renewal potential of CSCs with atypical activation of developmental signaling pathways involves the maintenance of stemness to support cancer progression. The acquisition of stemness in CSCs has been accomplished through genetic and epigenetic rewiring following the metabolic switch. In this context, "metabostemness" denotes the metabolic parameters that essentially govern the epitranscriptional gene reprogramming mechanism to dedifferentiate tumor cells into CSCs. Several metabolites often referred to as oncometabolites can directly remodel chromatin structure and thereby influence the operation of epitranscriptional circuits. This integrated metaboloepigenetic dimension of CSCs favors the differentiated cells to move in dedifferentiated macrostates. Some metabolic events might perform as early drivers of epitranscriptional reprogramming; however, subsequent metabolic hits may govern the retention of stemness properties in the tumor mass. Interestingly, selective removal of mitochondria through autophagy can promote metabolic plasticity and alter metabolic states during differentiation and dedifferentiation. In this connection, novel metabostemness-specific drugs can be generated as potential cancer therapeutics to target the metaboloepigenetic circuitry to eliminate CSCs.


Subject(s)
Mitophagy , Neoplasms , Cell Differentiation/physiology , Humans , Mitochondria/metabolism , Mitophagy/genetics , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism
4.
Int J Biochem Cell Biol ; 136: 106013, 2021 07.
Article in English | MEDLINE | ID: mdl-34022434

ABSTRACT

The NLR family pyrin domain containing 3 (NLRP3) inflammasome is responsible for the sensation of various pathogenic and non-pathogenic damage signals and has a vital role in neuroinflammation and neural diseases. Various stimuli, such as microbial infection, misfolded protein aggregates, and aberrant deposition of proteins can induce NLRP3 inflammasome in neural cells. Once triggered, the NLRP3 inflammasome leads to the activation of caspase-1, which in turn activates inflammatory cytokines, such as interleukin-1ß and interleukin -18, and induces pyroptotic cell death. Mitochondria are critically involved in diverse cellular processes and are involved in regulating cellular redox status, calcium levels, inflammasome activation, and cell death. Mitochondrial dysfunction and subsequent accumulation of mitochondrial reactive oxygen species, mitochondrial deoxyribonucleic acid, and other mitochondria-associated proteins and lipids play vital roles in the instigation of the NLRP3 inflammasome. In addition, the processes of mitochondrial dynamics, such as fission and fusion, are essential in the maintenance of mitochondrial integrity and their imbalance also promotes NLRP3 inflammasome activation. In this connection, mitophagy-mediated maintenance of mitochondrial homeostasis restricts NLRP3 inflammasome hyperactivation and its consequences in various neurological disorders. Hence, mitophagy can be exploited as a potential strategy to target damaged mitochondria induced NLRP3 inflammasome activation and its lethal consequences. Therefore, the identification of novel mitophagy modulators has promising therapeutic potential for NLRP3 inflammasome-associated neuronal diseases.


Subject(s)
Inflammasomes/metabolism , Inflammation/pathology , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mitophagy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Humans , Inflammation/etiology , Inflammation/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics
5.
Biochim Biophys Acta Rev Cancer ; 1876(1): 188561, 2021 08.
Article in English | MEDLINE | ID: mdl-33965511

ABSTRACT

Paired box 9 (PAX9) gene belongs to the PAX family, which encodes a family of metazoan transcription factors documented by a conserved DNA binding paired domain 128-amino-acids, critically essential for physiology and development. It is primarily expressed in embryonic tissues, such as the pharyngeal pouch endoderm, somites, neural crest-derived mesenchyme, and distal limb buds. PAX9 plays a vital role in craniofacial development by maintaining the odontogenic potential, mutations, and polymorphisms associated with the risk of tooth agenesis, hypodontia, and crown size in dentition. The loss-of-function of PAX9 in the murine model resulted in a short life span due to the arrest of cleft palate formation and skeletal abnormalities. According to recent studies, the PAX9 gene has a significant role in maintaining squamous cell differentiation, odontoblast differentiation of pluripotent stem cells, deregulation of which is associated with tumor initiation, and malignant transformation. Moreover, PAX9 contributes to promoter hypermethylation and alcohol- induced oro-esophageal squamous cell carcinoma mediated by downregulation of differentiation and apoptosis. Likewise, PAX9 activation is also reported to be associated with drug sensitivity. In summary, this current review aims to understand PAX9 function in the regulation of development, differentiation, and carcinogenesis, along with the underlying signaling pathways for possible cancer therapeutics.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , PAX9 Transcription Factor/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Drug Resistance, Neoplasm , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Genetic Therapy , Humans , Mutation , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Neoplastic Stem Cells/pathology , Organogenesis , PAX9 Transcription Factor/genetics , Polymorphism, Single Nucleotide , Signal Transduction
6.
J Mater Chem B ; 9(17): 3666-3676, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33949617

ABSTRACT

A novel hybrid drug carrier has been designed, taking N-doped mesoporous carbon (NMCS) as the core and PEG-PEI as the outer shell. NMCS was functionalized with a photocleavable nitrobenzyl-based linker following a click reaction. Gemcitabine was loaded into NMCS prior to the functionalization via π-π stacking interactions. NIR and the pH-responsive behavior of NMCS-linker-PEG-PEI bestow the multifunctional drug carrier with the controlled release of gemcitabine triggered by dual stimuli. The NMCS core upconverts NIR light to UV, which is absorbed by a photosensitive molecular gate and results in its cleavage and drug release. Further, NMCS converts NIR to heat, which deforms the outside polymer shell, thus triggering the drug release process. The release can be promptly arrested if the NIR source is switched off. A promising gemcitabine release of 75% has been achieved within 24 h under the dual stimuli of pH and temperature. NMCS-linker-PEG-PEI produced reactive oxygen species (ROS), which were verified in FaDu cells using flow cytometry. In vitro experiments showed that the NMCS-linker-PEG-PEI-GEM hybrid particle can induce synergistic therapeutic effects in FADU cells when exposed to the NIR light.


Subject(s)
Antineoplastic Agents/chemistry , Carbon/chemistry , Deoxycytidine/analogs & derivatives , Drug Carriers/chemistry , Nanospheres/chemistry , Photosensitizing Agents/chemistry , Polyethylene Glycols/chemistry , Polyethyleneimine/analogs & derivatives , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biocompatible Materials/chemistry , Cell Line, Tumor , Click Chemistry , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Drug Liberation , Humans , Hydrogen-Ion Concentration , Infrared Rays , Nitrobenzenes/chemistry , Oxidation-Reduction , Photochemotherapy , Photolysis , Photosensitizing Agents/pharmacology , Polyethyleneimine/chemistry , Porosity , Reactive Oxygen Species/metabolism , Surface Properties , Temperature , Time Factors , Gemcitabine
7.
Mitochondrion ; 57: 230-240, 2021 03.
Article in English | MEDLINE | ID: mdl-33476771

ABSTRACT

Mitochondrial quality control is crucial for sustaining cellular maintenance. Mitochondrial Ca2+ plays an important role in the maintenance of mitochondrial quality control through regulation of mitochondrial dynamics, mitophagy and mitochondrial biogenesis for preserving cellular homeostasis. The regulation of this dynamic interlink between these mitochondrial networks and mitochondrial Ca2+ appears indispensable for the adaptation of cells under external stimuli. Moreover, dysregulation of mitochondrial Ca2+ divulges impaired mitochondrial control that results in several pathological conditions such as cancer. Hence this review untangles the interplay between mitochondrial Ca2+ and quality control that govern mitochondrial health and mitochondrial coordinates in the development of cancer.


Subject(s)
Calcium Signaling , Mitochondria/metabolism , Neoplasms/metabolism , Calcium/metabolism , Cell Differentiation , Gene Expression Regulation, Neoplastic , Humans , Mitochondrial Dynamics
8.
Cancer Lett ; 498: 217-228, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33186655

ABSTRACT

Cancer stem cells (CSCs) are distinct subpopulations of cancer cells with stem cell-like abilities and are more resilient to chemotherapy, causing tumor relapse. Mitophagy, a selective form of autophagy, removes damaged unwanted mitochondria from cells through a lysosome-based degradation pathway to maintain cellular homeostasis. CSCs use mitophagy as a chief survival response mechanism for their growth, propagation, and tumorigenic ability. Mitochondrial biogenesis is a crucial cellular event replacing damaged mitochondria through the coordinated regulation of several transcription factors to achieve the bioenergetic demands of the cell. Because of the high mitochondrial content in CSCs, mitochondrial biogenesis is an interesting target to address the resistance mechanisms of anti-CSC therapy. However, to what extent both mitophagy and mitochondrial biogenesis are vital in promoting stemness, metabolic reprogramming, and drug resistance in CSCs has yet to be established. Therefore, in this review, we focus on understanding the interesting aspects of mitochondrial rewiring that involve mitophagy and mitochondrial biogenesis in CSCs. We also discuss their coordinated regulation in the elimination of CSCs, with respect to stemness and differentiation of the CSC phenotype, and the different aspects of tumorigenesis such as cancer initiation, progression, resistance, and tumor relapse. Finally, we address several other unanswered questions relating to targeted anti-CSC cancer therapy, which improves patient survival.


Subject(s)
Mitochondria/pathology , Mitochondria/physiology , Mitophagy/physiology , Neoplastic Stem Cells/pathology , Cell Differentiation/physiology , Drug Resistance, Neoplasm/physiology , Humans , Organelle Biogenesis
9.
Life Sci ; 264: 118722, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33160989

ABSTRACT

AIMS: Secretory clusterin (sCLU) plays an important role in tumor development and cancer progression. However, the molecular mechanisms and physiological functions of sCLU in oral cancer is unclear. We examined the impact of sCLU-mediated autophagy in cell survival and apoptosis inhibition in oral cancer. MAIN METHODS: Immunohistochemical analysis was performed to analyze protein expression in patient samples. Autophagy and mitophagy was studied by immunofluorescence microscopy and Western blot. The gain and loss of function was studied by overexpression of plasmid and siRNA approaches respectively. Cellular protection against nutrient starvation and therapeutic stress by sCLU was studied by cell viability, caspase assay and meta-analysis. KEY FINDINGS: The data from oral cancer patients showed that the expression levels of sCLU, ATG14, ULK1, and PARKIN increased in grade-wise manners. Interestingly, sCLU overexpression promoted autophagy through AMPK/Akt/mTOR signaling pathway leading to cell survival and protection from long exposure serum starvation induced-apoptosis. Additionally, sCLU was demonstrated to interact with ULK1 and inhibition of ULK1 activity by SBI206965 was found to abolish sCLU-induced autophagy indicating critical role of ULK1 in induction of autophagy. Furthermore, sCLU was observed to promote expression of mitophagy-associated proteins in serum starvation conditions to protect cells from nutrient deprivation. The meta-analysis elucidated that high CLU expression is associated with therapy resistance in cancer and we demonstrated that sCLU-mediated mitophagy was revealed to inhibit cell death by cisplatin. SIGNIFICANCE: The present investigation has highlighted the probable implications of the clusterin-induced autophagy in cell survival and inhibition of apoptosis in oral cancer.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy , Clusterin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mouth Neoplasms/pathology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Apoptosis/genetics , Autophagy/genetics , Cell Line, Tumor , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Mitophagy/genetics , Mouth Neoplasms/genetics , Neoplasms, Squamous Cell/genetics , Neoplasms, Squamous Cell/pathology
10.
Mol Biol Rep ; 47(9): 7209-7228, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32797349

ABSTRACT

Marine invertebrates are extremely diverse, largely productive, untapped oceanic resources with chemically unique bioactive lead compound contributing a wide range of screening for the discovery of anticancer compounds. The lead compounds have unfurled an extensive array of pharmacological properties owing to the presence of polyphenols, alkaloids, terpenoids and other secondary metabolites. The antioxidant, immunomodulatory and anti-tumor activities exhibited, are possibly regulated by the apoptosis induction, scavenging of ROS and modulation of cellular signaling pathways to defy the cellular deafness during carcinogenesis. Despite the enriched bioactive compounds, the marine invertebrates are largely unexplored as identification, screening, pre-clinical and clinical assessment of lead compounds and their synthetic analogs remain a major task to be solved. In the current review, we focus on the principle strategy and underlying mechanisms deployed by the bioactive anticancer compounds derived from marine invertebrates to combat cancer with special insight into the cell death mechanism.


Subject(s)
Antineoplastic Agents , Aquatic Organisms/chemistry , Invertebrates/chemistry , Neoplasms , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
11.
Free Radic Biol Med ; 160: 111-124, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32755671

ABSTRACT

Ionizing radiation has the potential to cause structural modification or change in electrochemical properties in parent lead pharmacophores that exhibit enhanced bioactivity. Gallic acid (GA), a triphenolic compound has displayed potent anticancer drug potency due to its withstanding antioxidant propensity. This study uncovered the comparative efficacy of gamma-irradiated gallic acid (GAIR) in the modulation of an antioxidant system for regulation apoptosis and autophagy. GAIR exhibited remarkable anti-proliferative efficacy as shown by MTT, clonogenic survival, and scratch assay. In addition to this, GAIR promoted intrinsic apoptosis through mitochondrial superoxide generation. GAIR decreased the activity of antioxidant enzymes by downregulating nuclear factor erythroid 2-related factor 2 (NRF2) and its downstream effector molecules NAD(P)H Quinone Dehydrogenase 1 (NQO1) and gamma-glutamylcysteine synthetase (GCLC). Simultaneously, GAIR attenuated autophagosome-lysosome fusion without altering the lysosomal activity. Inhibition of autophagic flux resulted in the accumulation of lipid droplets (LDs) such as hexadecanoic acid and oleic acid that fuelled superoxide generation leading to apoptosis. In the meantime, under oxidative upset, conversion of LDs to free fatty acids reduced leading to inhibition of ATP generation that subsequently provoked apoptosis. The effects of autophagy inhibition by GAIR on the therapeutic efficacy of chemotherapeutic drugs was studied and the co-treatment markedly decreased the cell viability and increased apoptosis. Further, GAIR exhibited potent antitumor activity in Dalton's Lymphoma-tumor bearing mice through modulation of apoptosis and autophagy without toxic activity. In conclusion, change in electrochemical properties by gamma radiation enhances the anticancer efficacy of gallic acid through superoxide mediated apoptosis fuelled by inhibition of lipophagy in an NRF2 dependent signaling pathway.


Subject(s)
Apoptosis , Autophagy , Gallic Acid , NF-E2-Related Factor 2 , Animals , Gallic Acid/pharmacology , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species , Signal Transduction
12.
Food Chem Toxicol ; 136: 111073, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31877368

ABSTRACT

Terminalia bellirica (TB) has been used in traditional Indian medical system, Ayurveda. However, the mechanism underlying the efficacy of the TB extract against oral squamous cell carcinoma (OSCC) is yet to be explored. The present study established a connecting link between the TB extract induced apoptosis and autophagy in relation to reactive oxygen species (ROS). Our study revealed, that gallic acid in the TB extract possess a strong free radical scavenging capacity contributing towards the selective anti-proliferative activity. Furthermore, TB extract markedly enhanced the accumulation of ROS that facilitated mitochondrial apoptosis through DNA damage, indicating ROS as the vital component in regulation of apoptosis. This effect was effectively reversed by the use of a ROS scavenger, N-acetyl cysteine (NAC). Moreover, it was observed to induce autophagy; however, it attenuated the autophagosome-lysosome fusion in Cal33 cells without altering the lysosomal activity. Pharmacological inhibitors of autophagy, namely, 3-methyladenine and chloroquine, were demonstarated to regulate the stage-specific progression of autophagy post treatment with the TB extract, favouring subsequent activation of apoptosis. These findings revealed, presence of gallic acid in TB extract below NOAEL value causes oxidative upset in oral cancer cells and promote programmed cell death which has a potential therapeutic value against oral squamous cell carcinoma.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Mouth Neoplasms/physiopathology , Plant Extracts/pharmacology , Terminalia/chemistry , Antineoplastic Agents, Alkylating/analysis , Carcinoma, Squamous Cell , Cell Line, Tumor , DNA Damage/drug effects , Humans , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Plant Extracts/analysis , Reactive Oxygen Species/metabolism
13.
Langmuir ; 35(47): 15320-15329, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31682135

ABSTRACT

Use of nanomaterials blessed with both therapeutic and diagnostic properties is a proficient strategy in the treatment of cancer in its early stage. In this context, our paper reports the synthesis of uniform size N-rich mesoporous carbon nanospheres of size 65-70 nm from pyrrole and aniline precursors using Triton-X as a structure-directing agent. Transmission electron microscopy reveals that these carbons spheres contain void spaces in which ultrasmall nitrogen-doped quantum dots (NCQD) are captured within the matrix. These mesoporous hollow NCQD captured carbon spheres (NCQD-HCS) show fluorescence quantum yield up to 14.6% under λex = 340 nm. Interestingly, samples calcined at >800 °C clearly absorb in the wavelength range 700-1000 nm and shows light-to-heat conversion efficiency up to 52%. In vitro experiments in human oral cancer cells (FaDu) show that NCQD-HCS are internalized by the cells and induce a substantial thermal ablation effect in FaDu cells when exposed under a 980 nm near-infrared laser.


Subject(s)
Antineoplastic Agents/pharmacology , Fluorescent Dyes/pharmacology , Quantum Dots/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Apoptosis/drug effects , Carbon/chemistry , Carbon/radiation effects , Cell Line, Tumor , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/radiation effects , Humans , Hyperthermia, Induced/methods , Infrared Rays , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Mouth Neoplasms/drug therapy , Nitrogen/chemistry , Nitrogen/radiation effects , Phototherapy/methods , Quantum Dots/radiation effects , Theranostic Nanomedicine/methods
15.
Life Sci ; 111(1-2): 27-35, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25064824

ABSTRACT

AIMS: The present study evaluated the potential role of soybean lectin's (SBL) anticancer effect in vitro in different cancer cell lines and the therapeutic effectiveness in vivo in Dalton's lymphoma (DL) bearing mice model. MAIN METHODS: The effect of SBL on cell growth and viability was measured using MTT assay in different cancer cells in vitro. Apoptosis, autophagic cell death, DNA-damaging potential and reactive oxygen species (ROS) were analyzed in HeLa cells. The in vivo efficacy of SBL was demonstrated in Dalton's lymphoma (DL) bearing mice. KEY FINDINGS: SBL demonstrated clear, strong antiproliferative activity without affecting normal cells; however, heat denaturation of SBL diminished the antiproliferative efficacy of molecule as demonstrated by MTT assay. A sharp 74.51 ± 3.5% and 82.95 ± 5.8% inhibition of tumor cell proliferation in DL mice occurred when SBL was administered at a dosage of 1 and 2mg/kg body weight (i.p.), respectively, for ten days with the induction of autophagic and apoptotic cell death. An in vitro investigation revealed that SBL-mediated autophagy, apoptosis and DNA damage in HeLa cells were inflicted through the generation of ROS in a dose-dependent manner. Interestingly, pre-treating HeLa cells with N-acetylcysteine (NAC), a typical ROS scavenger, led to a noticeable reduction in SBL-induced autophagy, apoptosis and DNA-damaging activities, suggesting that SBL's antitumor potential was governed by ROS activation. SIGNIFICANCE: In this study, we evaluated the apoptotic, autophagic death, and DNA-damaging effects of SBL in cancer cells, which may have the potential to be used as a phyto-derived protein for cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Neoplasms/drug therapy , Plant Lectins/pharmacology , Reactive Oxygen Species/metabolism , Soybean Proteins/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , HeLa Cells/drug effects , Hep G2 Cells/drug effects , Humans , In Vitro Techniques , Lymphoma/drug therapy , Mice , Neoplasms, Experimental/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...